1.Pathophysiological implications of cellular senescence and prospects for novel anti-aging drugs.
Acta Physiologica Sinica 2023;75(6):847-863
Chronological aging is the leading risk factor for human diseases, while aging at the cellular level, namely cellular senescence, is the fundamental driving force of organismal aging. The impact of cellular senescence on various life processes, including normal physiology, organismal aging and the progress of various age-related pathologies, has been largely ignored for a long time. However, with recent advancement in relevant fields, cellular senescence has become the core of aging biology and geriatric medicine. Although senescent cells play important roles in physiological processes including tissue repair, wound healing, and embryonic development, they can also contribute to tissue dysfunction, organ degeneration and various pathological conditions during adulthood. Senescent cells exert paracrine effects on neighboring cells in tissue microenvironments by developing a senescence-associated secretory phenotype, thus maintaining long-term and active intercellular communications that ultimately results in multiple pathophysiological effects. This is regarded as one of the most important discoveries in life science of this century. Notably, selective elimination of senescent cells through inducing their apoptosis or specifically inhibiting the senescence-associated secretory phenotype has shown remarkable potential in preclinical and clinical interventions of aging and age-related diseases. This reinforces the belief that senescent cells are the key drug target to alleviate various aging syndromes. However, senescent cells exhibit heterogeneity in terms of form, function and tissue distribution, and even differ among species, which presents a challenge for the translation of significant research achievements to clinical practice in future. This article reviews and discusses the characteristics of senescent cells, current targeting strategies and future trends, providing useful and valuable references for the rapidly blooming aging biology and geriatric medicine.
Humans
;
Adult
;
Aged
;
Cellular Senescence/genetics*
;
Aging
;
Apoptosis
;
Cell Communication
;
Wound Healing/physiology*
2.Single-cell transcriptomics reveals gene signatures and alterations associated with aging in distinct neural stem/progenitor cell subpopulations.
Zhanping SHI ; Yanan GENG ; Jiping LIU ; Huina ZHANG ; Liqiang ZHOU ; Quan LIN ; Juehua YU ; Kunshan ZHANG ; Jie LIU ; Xinpei GAO ; Chunxue ZHANG ; Yinan YAO ; Chong ZHANG ; Yi E SUN
Protein & Cell 2018;9(4):351-364
Aging associated cognitive decline has been linked to dampened neural stem/progenitor cells (NSC/NPCs) activities manifested by decreased proliferation, reduced propensity to produce neurons, and increased differentiation into astrocytes. While gene transcription changes objectively reveal molecular alterations of cells undergoing various biological processes, the search for molecular mechanisms underlying aging of NSC/NPCs has been confronted by the enormous heterogeneity in cellular compositions of the brain and the complex cellular microenvironment where NSC/NPCs reside. Moreover, brain NSC/NPCs themselves are not a homogenous population, making it even more difficult to uncover NSC/NPC sub-type specific aging mechanisms. Here, using both population-based and single cell transcriptome analyses of young and aged mouse forebrain ependymal and subependymal regions and comprehensive "big-data" processing, we report that NSC/NPCs reside in a rather inflammatory environment in aged brain, which likely contributes to the differentiation bias towards astrocytes versus neurons. Moreover, single cell transcriptome analyses revealed that different aged NSC/NPC subpopulations, while all have reduced cell proliferation, use different gene transcription programs to regulate age-dependent decline in cell cycle. Interestingly, changes in cell proliferation capacity are not influenced by inflammatory cytokines, but likely result from cell intrinsic mechanisms. The Erk/Mapk pathway appears to be critically involved in regulating age-dependent changes in the capacity for NSC/NPCs to undergo clonal expansion. Together this study is the first example of using population and single cell based transcriptome analyses to unveil the molecular interplay between different NSC/NPCs and their microenvironment in the context of the aging brain.
Aging
;
genetics
;
Animals
;
Astrocytes
;
cytology
;
metabolism
;
Brain
;
cytology
;
metabolism
;
Cell Differentiation
;
genetics
;
Cell Division
;
genetics
;
Cell Proliferation
;
genetics
;
Gene Expression Regulation
;
genetics
;
Mice
;
Neural Stem Cells
;
metabolism
;
Single-Cell Analysis
;
Stem Cells
;
cytology
;
metabolism
;
Transcriptome
;
genetics
3.Decreased expression of DNA repair proteins Ku70 and Mre11 is associated with aging and may contribute to the cellular senescence.
Yeun Jin JU ; Kee Ho LEE ; Jeong Eun PARK ; Yong Su YI ; Mi Yong YUN ; Yong Ho HAM ; Tae Jin KIM ; Hyun Mi CHOI ; Gwi Jung HAN ; Jong Hoon LEE ; Juneyoung LEE ; Jong Seol HAN ; Kyung Mi LEE ; Gil Hong PARK
Experimental & Molecular Medicine 2006;38(6):686-693
The gradual loss of telomeric DNA can contribute to replicative senescence and thus, having longer telomeric DNA is generally considered to provide a longer lifespan. Maintenance and stabilization of telomeric DNA is assisted by binding of multiple DNA-binding proteins, including those involved in double strand break (DSB) repair. We reasoned that declining DSB repair capacity and increased telomere shortening in aged individuals may be associated with decreased expression of DSB repair proteins capable of telomere binding. Our data presented here show that among the DSB repair proteins tested, only the expression of Ku70 and Mre11 showed statistically significant age-dependent changes in human lymphocytes. Furthermore, we found that expressions of Ku70 and Mre11 are statistically correlated, which indicate that the function of Ku70 and Mre11 may be related. All the other DSB repair proteins tested, Sir2, TRF1 and Ku80, did not show any significant differences upon aging. In line with these data, people who live in the regional community (longevity group), which was found to have statistically longer average life span than the rest area, shows higher level of Ku70 expression than those living in the neighboring control community. Taken together, our data show, for the first time, that Ku70 and Mre11 may represent new biomarkers for aging and further suggest that maintenance of higher expression of Ku70 and Mre11 may be responsible for keeping longer life span observed in the longevity group.
Telomere/genetics
;
Middle Aged
;
Longevity
;
Humans
;
DNA-Binding Proteins/*metabolism
;
DNA Repair/*genetics
;
DNA/genetics
;
Cell Aging/*physiology
;
CD4-Positive T-Lymphocytes/metabolism
;
Antigens, Nuclear/*metabolism
;
Aging/*physiology
;
Aged, 80 and over
;
Aged
;
Adult
4.Loss of KDM4B impairs osteogenic differentiation of OMSCs and promotes oral bone aging.
Peng DENG ; Insoon CHANG ; Jiongke WANG ; Amr A BADRELDIN ; Xiyao LI ; Bo YU ; Cun-Yu WANG
International Journal of Oral Science 2022;14(1):24-24
Aging of craniofacial skeleton significantly impairs the repair and regeneration of trauma-induced bony defects, and complicates dental treatment outcomes. Age-related alveolar bone loss could be attributed to decreased progenitor pool through senescence, imbalance in bone metabolism and bone-fat ratio. Mesenchymal stem cells isolated from oral bones (OMSCs) have distinct lineage propensities and characteristics compared to MSCs from long bones, and are more suited for craniofacial regeneration. However, the effect of epigenetic modifications regulating OMSC differentiation and senescence in aging has not yet been investigated. In this study, we found that the histone demethylase KDM4B plays an essential role in regulating the osteogenesis of OMSCs and oral bone aging. Loss of KDM4B in OMSCs leads to inhibition of osteogenesis. Moreover, KDM4B loss promoted adipogenesis and OMSC senescence which further impairs bone-fat balance in the mandible. Together, our data suggest that KDM4B may underpin the molecular mechanisms of OMSC fate determination and alveolar bone homeostasis in skeletal aging, and present as a promising therapeutic target for addressing craniofacial skeletal defects associated with age-related deteriorations.
Aging
;
Cell Differentiation
;
Facial Bones/physiology*
;
Humans
;
Jumonji Domain-Containing Histone Demethylases/genetics*
;
Mesenchymal Stem Cells/cytology*
;
Osteogenesis
;
Osteoporosis
5.Effects of Aging on the Proliferation and Differentiation Capacity of Human Periodontal Ligament Stem Cells.
Tingting DU ; Na LIU ; Bin GU ; Ying LI ; Yifang YUAN ; Wei ZHANG ; Tong ZHANG
Chinese Medical Sciences Journal 2017;32(2):83-81
Objective The aim of this study is to investigate the proliferation, differentiation and apoptosis of periodontal ligament stem cells (PDLSC) derived from different aged donors, and to evaluate the effects of aging on the biological characteristics of PDLSC.Methods Periodontal ligament tissues were obtained from 24 surgically extracted human premolars during orthodontics therapy. The specimens were divided into three groups according to the donor's age. Group A: 18-20 years, group B: 30-35 years, group C: 45-50 years. PDLSC were isolated and cultured using a tissue-block-based enzymolytic method by limiting dilution assay. The colony forming efficiency of PDLSC for three experimental groups was determined. Senescence-Associated β-Galactosidase (SA-β-G) expression in the three groups was examined using β-galactosidase staining working solution. Cell cycle and apoptosis of the PDLSC were examined by the flow cytometry. Alkaline phosphatase (ALP) activity was evaluated by ALP staining. The expression of osteoplastic differentiation related genes Runt-related transcription factor-2 (Runx-2), Collagen Type 1 (col-1), and ALP of PDLSC were examined by quantitative real-time RT-PCR.Results The colony forming efficiency of PDLSC in Group A, B and C was 36.67%, 22.67% and 9.33%, respectively, which decreased with donors' age (P<0.05). SA-β-G expression of the senescent PDLSC in group A, B and C were 4.14%, 16.39%, 50.38%, respectively (P<0.05). Cells in G2/S phase was 38.73%, 29.88%, 18.25% (P<0.05), and the apoptosis rate was 1.57%, 4.56%, 5.84% (P<0.05), in group A, B and C respectively. The ALP staining in the three groups decreased with the increase of donors' ages, and the expression of Runx-2, col-1 and ALP decreased gradually from group A to group C (all P<0.05), which indicated the osteogenic differentiation capacity of PDLSC decreased while donor aging.Conclusion Human PDLSC could be successfully isolated from periodontal ligament tissues of different aged donors. However, the proliferation and osteogenic differentiation capacity of PDLSC decreased while donor aging.
Adolescent
;
Adult
;
Aging
;
Alkaline Phosphatase
;
genetics
;
Apoptosis
;
Cell Cycle
;
Cell Differentiation
;
Cell Proliferation
;
Humans
;
Middle Aged
;
Osteogenesis
;
Periodontal Ligament
;
cytology
;
Stem Cells
;
cytology
6.p41-Arc, a regulatory subunit of Arp2/3 complex, can induce premature senescence in the absence of p53 and Rb.
Un Jung YUN ; Sang Eun PARK ; Deug Y SHIN
Experimental & Molecular Medicine 2011;43(7):389-392
Cellular senescence is a tumor-suppressive process instigated by proliferation in the absence of telomere replication, by cellular stresses such as oncogene activation, or by activation of the tumor suppressor proteins, such as Rb or p53. This process is characterized by an irreversible cell cycle exit, a unique morphology, and expression of senescence-associated-beta-galactosidase (SA-beta-gal). Despite the potential biological importance of cellular senescence, little is known of the mechanisms leading to the senescent phenotype. p41-Arc has been known to be a putative regulatory component of the mammalian Arp2/3 complex, which is required for the formation of branched networks of actin filaments at the cell cortex. In this study, we demonstrate that p41-Arc can induce senescent phenotypes when it is overexpressed in human tumor cell line, SaOs-2, which is deficient in p53 and Rb tumor suppressor genes, implying that p41 can induce senescence in a p53-independent way. p41-Arc overexpression causes a change in actin filaments, accumulating actin filaments in nuclei. Therefore, these results imply that a change in actin filament can trigger an intrinsic senescence program in the absence of p53 and Rb tumor suppressor genes.
Actin Cytoskeleton/metabolism
;
Actin-Related Protein 2-3 Complex/*metabolism
;
*Cell Aging
;
Cell Cycle Proteins/metabolism
;
Cell Line, Tumor
;
Cell Nucleus/metabolism
;
Fibroblasts/physiology
;
Humans
;
Recombinant Proteins/genetics/*metabolism
;
Retinoblastoma Protein/*deficiency/genetics
;
Tumor Suppressor Protein p53/*deficiency/genetics
7.Immortalization of human embryonic fibroblasts by overexpression of c-myc and simian virus 40 large T antigen.
Hyun Seok KIM ; Jong Yeon SHIN ; Ji Yeon YUN ; Duck Kyu AHN ; Jae Yong LEE
Experimental & Molecular Medicine 2001;33(4):293-298
SV40 large T antigen, a viral oncoprotein, is known to immortalize human diploid fibroblast by soaking up cellular RB and p53, but its frequency is extremely low. Additional genetic alteration is necessary for single-step immortalization. We attempted to find out what this alteration is by overexpressing cellular signal mediator genes; c-myc and cyclin D frequently amplified in many cancer cells. Overexpression of cyclin D did not affect the immortalization, but, overexpression of c-myc along with T antigen could immortalize normal human diploid fibroblast. Several cellular markers tested during immortalization process showed that p21, a cyclin-dependent kinase inhibitor and a marker of cellular senescence, disappeared in the life span-extended cells by T antigen and in the immortalized cells by c-myc. p21 was, however, elevated in the senescent cells and in the cells of crisis. Interestingly, p16 was upregulated whenever T antigen is overexpressed. Telomerase activity was also activated only in the immortalized cells. These results suggest that overexpression of c-myc contributes to immortalization of human diploid fibroblast by activating telomerase activity and suppressing p21 activity.
Antigens, Polyomavirus Transforming/genetics/*metabolism
;
Biological Markers
;
Cell Aging/*genetics
;
Cell Transformation, Viral
;
Cells, Cultured
;
Cyclins/metabolism
;
Diploidy
;
Fibroblasts/*metabolism
;
Genes, myc/*genetics
;
Human
;
Protein p16/metabolism
;
Simian virus 40/genetics
;
Support, Non-U.S. Gov't
;
Telomerase/metabolism
8.Immortalization of human embryonic fibroblasts by overexpression of c-myc and simian virus 40 large T antigen.
Hyun Seok KIM ; Jong Yeon SHIN ; Ji Yeon YUN ; Duck Kyu AHN ; Jae Yong LEE
Experimental & Molecular Medicine 2001;33(4):293-298
SV40 large T antigen, a viral oncoprotein, is known to immortalize human diploid fibroblast by soaking up cellular RB and p53, but its frequency is extremely low. Additional genetic alteration is necessary for single-step immortalization. We attempted to find out what this alteration is by overexpressing cellular signal mediator genes; c-myc and cyclin D frequently amplified in many cancer cells. Overexpression of cyclin D did not affect the immortalization, but, overexpression of c-myc along with T antigen could immortalize normal human diploid fibroblast. Several cellular markers tested during immortalization process showed that p21, a cyclin-dependent kinase inhibitor and a marker of cellular senescence, disappeared in the life span-extended cells by T antigen and in the immortalized cells by c-myc. p21 was, however, elevated in the senescent cells and in the cells of crisis. Interestingly, p16 was upregulated whenever T antigen is overexpressed. Telomerase activity was also activated only in the immortalized cells. These results suggest that overexpression of c-myc contributes to immortalization of human diploid fibroblast by activating telomerase activity and suppressing p21 activity.
Antigens, Polyomavirus Transforming/genetics/*metabolism
;
Biological Markers
;
Cell Aging/*genetics
;
Cell Transformation, Viral
;
Cells, Cultured
;
Cyclins/metabolism
;
Diploidy
;
Fibroblasts/*metabolism
;
Genes, myc/*genetics
;
Human
;
Protein p16/metabolism
;
Simian virus 40/genetics
;
Support, Non-U.S. Gov't
;
Telomerase/metabolism
9.Experimental study on aging effect of Angelica sinensis polysaccharides combined with cytarabine on human leukemia KG1alpha cell lines.
Chun-Yan XU ; Shan GENG ; Jun LIU ; Jia-Hong ZHU ; Xian-Ping ZHANG ; Rong JIANG ; Ya-Ping WANG
China Journal of Chinese Materia Medica 2014;39(7):1260-1264
The latest findings of our laboratory showed that Angelica sinensis polysaccharide (ASP) showed a definite effect in regulating the aging of hematopoietic stem cells. Leukemia is a type of malignant hematopoietic tumor in hematopoietic stem cells. There have been no relevant reports about ASP's effect in regulating the aging of leukemia cells. In this study, human acute myeloid leukemia (AML) KG1alpha cell lines in logarithmic growth phase were taken as the study object, and were divided into the ASP group, the cytarabine (Ara-C) group, the ASP + Ara-C group and the control group. The groups were respectively treated with different concentration of ASP, Ara-C and ASP + Ara-C for different periods, with the aim to study the effect of ASP combined with Ara-C in regulating the aging of human acute myeloid leukemia KG1alpha cell lines and its relevant mechanism. The results showed that ASP, Ara-C and ASP + Ara-C could obviously inhibit KG1alpha cell proliferation in vitro, block the cells in G0/G1 phase. The cells showed the aging morphological feature. The percentage of positive stained aging cells was dramatically increased, and could significantly up-regulate the expression of aging-related proteins P16 and RB, which were more obvious in the ASP + Ara-C group. In conclusion, the aging mechanism of KG1alpha cell induced by ASP and Ara-C may be related to the regulation of the expression of aging-related proteins, suggesting that the combined administration of ASP and anticancer drugs plays a better role in the treatment of leukemia .
Aging
;
drug effects
;
genetics
;
metabolism
;
Angelica sinensis
;
chemistry
;
Cell Cycle
;
drug effects
;
Cell Proliferation
;
drug effects
;
Cyclin-Dependent Kinase Inhibitor p16
;
genetics
;
metabolism
;
Humans
;
Leukemia
;
drug therapy
;
genetics
;
metabolism
;
physiopathology
;
Polysaccharides
;
pharmacology
;
Retinoblastoma Protein
;
genetics
;
metabolism
;
Tumor Cells, Cultured
10.Proceedings of human mitochondrial DNA study and its applications to forensic medicine.
Xue-Ping ZHOU ; Wei-Juan ZHANG ; Zhen-Jun JIA
Journal of Forensic Medicine 2004;20(2):113-119
The mitochondrial DNA (mtDNA) is a small circular genome located within the mitochondria in the cytoplasm of the cell. Evidence of its existence first arose over 30 years ago. Now the field of the mitochondria is one of the fastest growing disciplines in biomedicine which is driven by fundamentally interesting questions. These questions are mainly about the way of mitochondria evolving and energy producing. In addition, what the consequences of mitochondrial genome mutations in diseases are? How program cell death is regulated? What happens to mitochondria when aging? These questions remain to be answered and the basic understanding of them will contribute to anthropological and forensic analysis, as well as therapy of many diseases. The following review has brought this question to notice by summarizing recent mitochondria research.
Aging/genetics*
;
Apoptosis
;
Cell Nucleus/genetics*
;
DNA, Mitochondrial/genetics*
;
Forensic Medicine
;
Genome, Human/genetics*
;
Humans
;
Mitochondrial Diseases/prevention & control*
;
Molecular Sequence Data
;
Mutation
;
Oligonucleotide Probes
;
Polymorphism, Single-Stranded Conformational
;
Sequence Analysis, DNA